The st7 protein is a homolog of the human ST7 gene, which encodes a transmembrane receptor-like protein. In zebrafish, it is part of the LDLR superfamily, a group of proteins involved in lipid metabolism, signal transduction, and cellular uptake. The recombinant form is produced via heterologous expression in host systems such as E. coli, yeast, or mammalian cells .
Key Properties | Details |
---|---|
Gene Names | st7, zgc:136822 |
UniProt ID | Q1RLU8 |
Molecular Weight | ~58,607 Da (full-length) |
Function | Signal transduction, endocytosis, potential tumor suppression |
Recombinant st7 is synthesized using diverse expression systems, with purity and stability optimized for research applications:
st7 interacts with proteins involved in cellular signaling and trafficking:
Interacting Partners | Function | Source |
---|---|---|
RACK1 | Signal transduction, receptor-mediated processes | |
MIBP | Integrin signaling, cell adhesion | |
SARA | SMAD anchor, TGF-β signaling |
Tumor Suppression:
Human ST7 regulates genes like SERPINE1 (cellular structure) and Survivin (apoptosis), suggesting a role in oncogenic pathways . While direct evidence in zebrafish is limited, homology to human ST7 implies conserved mechanisms.
Recombinant st7 is used to produce polyclonal antibodies for:
Recombinant st7 aids in studying:
Signal Transduction: Interactions with RACK1, MIBP, and SARA .
Toxicology: Protein profiling in zebrafish embryos exposed to pollutants (e.g., PFOS) .
KEGG: dre:678587
UniGene: Dr.43903
Multiple expression systems have been validated for zebrafish ST7 protein production. According to available data, recombinant Danio rerio ST7 can be effectively expressed in:
Cell-free expression systems: Offering rapid production with reduced complexity for initial characterization studies .
E. coli expression systems: Providing high yield though potentially lacking some post-translational modifications .
Baculovirus expression: Suitable for producing ST7 with more complex folding requirements .
Mammalian cell expression: Offering the most physiologically relevant post-translational modifications .
The selection depends on experimental requirements regarding protein yield, purity, and post-translational modifications. For structural studies requiring high purity, E. coli systems followed by SDS-PAGE validation to confirm ≥85% purity is recommended .
Verification protocols should include multiple complementary techniques:
SDS-PAGE analysis: Should demonstrate ≥85% purity with the expected molecular weight band .
Western blot analysis: Using validated antibodies such as rabbit anti-Danio rerio ST7 polyclonal antibodies for specific detection .
Mass spectrometry: For peptide fingerprinting to confirm protein identity.
Functional assays: To verify biological activity of the purified protein.
For optimal results, purified protein should be aliquoted, flash-frozen, and stored at -80°C to maintain stability and prevent freeze-thaw cycles.
Several approaches can be employed to generate ST7-deficient zebrafish models:
Zinc Finger Nucleases (ZFNs): These can be designed to target specific sites within the st7 gene. The approach involves:
In silico analysis of the st7 cDNA sequence to identify potential zinc finger binding sites
Assembly of 4-finger ZFPs directed against distinct positions in the st7 locus
Validation of ZFP binding affinity using ELISA assays
Testing ZFN activity in a yeast-based system
Microinjection of validated ZFN mRNAs into 1-cell zebrafish embryos
Screening for mutations using PCR and restriction enzyme digestion
CRISPR/Cas9 system: This more recent technique offers improved efficiency and specificity:
Design of guide RNAs targeting conserved exons of the st7 gene
Co-injection of guide RNAs and Cas9 mRNA into zebrafish embryos
Validation of mutations through sequencing and phenotypic analysis
Both approaches typically yield small insertions or deletions at the target site, which can disrupt gene function when appropriately positioned .
When using genome editing tools such as ZFNs or CRISPR/Cas9 to target the st7 gene, off-target assessment is critical:
In silico prediction:
Experimental validation:
Direct analysis of predicted off-target sites by PCR amplification and sequencing
Whole genome sequencing of selected mutant lines to identify unpredicted off-target effects
Complementation testing by crossing different mutant lines to confirm phenotypes are due to st7 disruption rather than off-target effects
Phenotypic validation:
Perform rescue experiments by introducing wild-type st7 to confirm phenotype specificity
Compare multiple independently generated mutant lines to confirm consistent phenotypes
This multi-layered approach ensures that observed phenotypes are specifically related to st7 disruption rather than off-target effects .
To comprehensively investigate ST7 function in zebrafish, consider these methodological approaches:
Spatiotemporal expression profiling:
Whole-mount in situ hybridization at different developmental stages
Transgenic reporter lines with fluorescent proteins under st7 promoter control
Quantitative PCR to measure expression levels across tissues and timepoints
Loss-of-function studies:
Analysis of morphological phenotypes in st7 mutants/knockdowns
Histological examination of affected tissues
Behavioral assays to assess functional outcomes
Disease model integration:
These approaches should be applied within the developmental window where zebrafish rely exclusively on innate immune responses (up to 4-6 weeks post-fertilization), providing an opportunity to examine st7 function without the confounding effects of adaptive immunity .
Multiple complementary approaches can identify protein-protein interactions involving ST7:
Co-immunoprecipitation (Co-IP):
Using anti-ST7 antibodies to pull down protein complexes from zebrafish tissue lysates
Mass spectrometry analysis of co-precipitated proteins
Validation of identified interactions using reverse Co-IP
Proximity labeling approaches:
Generation of ST7-BioID or ST7-APEX2 fusion proteins
Expression in zebrafish through transgenesis or mRNA injection
Identification of proximal proteins through streptavidin pulldown and mass spectrometry
Yeast two-hybrid screening:
Using ST7 as bait against a zebrafish cDNA library
Validation of positive interactions through secondary assays
In vivo confirmation in zebrafish embryos
Each method has distinct advantages and limitations, so combining multiple approaches provides the most robust identification of physiologically relevant interactions.
For comprehensive ST7 expression analysis in zebrafish tissues:
At the mRNA level:
Quantitative RT-PCR: Provides precise quantification across different tissues
In situ hybridization: Reveals spatial distribution within tissues
RNA-seq: Offers genome-wide context for st7 expression patterns
At the protein level:
For real-time visualization:
Transgenic reporter lines expressing fluorescent proteins under st7 promoter control
Time-lapse imaging during developmental processes
For developmental studies, stage-dependent expression analysis should be performed systematically from embryonic development through larval stages to maturity, as expression patterns may change significantly across development .
For transcriptomic analysis of ST7-regulated pathways:
Experimental design recommendations:
Compare wild-type vs. st7 mutant/knockdown zebrafish at multiple developmental timepoints
Include biological replicates (minimum n=3 for each condition)
Consider tissue-specific RNA isolation for targeted analysis
Bioinformatic analysis workflow:
Differential gene expression analysis using DESeq2 or similar tools
Pathway enrichment analysis using KEGG, GO, and Reactome databases
Upstream regulator prediction using tools like IPA or LISA
Validation approach:
qRT-PCR confirmation of key differentially expressed genes
Protein-level validation of selected targets
Functional studies of identified pathways using pharmacological inhibitors or genetic approaches
This comprehensive approach allows for the identification of both direct and indirect targets of ST7 regulation, providing insights into its biological function.
Several methodological challenges exist when characterizing post-translational modifications (PTMs) of recombinant ST7:
Expression system selection:
Analytical approaches for PTM characterization:
Mass spectrometry protocols for phosphorylation site mapping
Glycan analysis through lectin affinity or specialized MS approaches
Site-directed mutagenesis to determine functional significance of identified PTMs
Comparative analysis recommendations:
Parallel characterization of native ST7 purified from zebrafish tissues
Comparison across expression systems to identify critical modifications
Functional assays to determine the impact of specific PTMs
These challenges underscore the importance of selecting appropriate expression systems based on experimental requirements for studying ST7 function and structure.
Recombinant proteins often present solubility and stability challenges. For ST7:
Solubility enhancement strategies:
Fusion tags optimization (SUMO, MBP, or GST tags often improve solubility)
Expression temperature and induction optimization
Co-expression with molecular chaperones
Buffer screening using high-throughput approaches
Stability assessment and improvement:
Differential scanning fluorimetry to identify stabilizing conditions
Addition of specific cofactors or binding partners
Identification of minimal stable domains through limited proteolysis
Storage condition optimization (cryoprotectants, pH, ionic strength)
Quality control protocols:
Regular assessment of protein activity over time
Size-exclusion chromatography to monitor aggregation
Circular dichroism to evaluate secondary structure maintenance
These approaches should be systematically tested and optimized for the specific construct and expression system used for ST7 production.